Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.668
Filtrar
1.
Molecules ; 29(9)2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38731628

RESUMEN

Fluorescence lifetime imaging microscopy (FLIM) has proven to be a useful method for analyzing various aspects of material science and biology, like the supramolecular organization of (slightly) fluorescent compounds or the metabolic activity in non-labeled cells; in particular, FLIM phasor analysis (phasor-FLIM) has the potential for an intuitive representation of complex fluorescence decays and therefore of the analyzed properties. Here we present and make available tools to fully exploit this potential, in particular by coding via hue, saturation, and intensity the phasor positions and their weights both in the phasor plot and in the microscope image. We apply these tools to analyze FLIM data acquired via two-photon microscopy to visualize: (i) different phases of the drug pioglitazone (PGZ) in solutions and/or crystals, (ii) the position in the phasor plot of non-labelled poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), and (iii) the effect of PGZ or PGZ-containing NPs on the metabolism of insulinoma (INS-1 E) model cells. PGZ is recognized for its efficacy in addressing insulin resistance and hyperglycemia in type 2 diabetes mellitus, and polymeric nanoparticles offer versatile platforms for drug delivery due to their biocompatibility and controlled release kinetics. This study lays the foundation for a better understanding via phasor-FLIM of the organization and effects of drugs, in particular, PGZ, within NPs, aiming at better control of encapsulation and pharmacokinetics, and potentially at novel anti-diabetics theragnostic nanotools.


Asunto(s)
Nanopartículas , Pioglitazona , Pioglitazona/farmacología , Pioglitazona/química , Nanopartículas/química , Animales , Línea Celular Tumoral , Humanos , Microscopía Fluorescente/métodos , Ratas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Hipoglucemiantes/farmacología , Hipoglucemiantes/química
2.
Bull Exp Biol Med ; 176(5): 697-702, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38724814

RESUMEN

One of the key problems of glioblastoma treatment is the low effectiveness of chemotherapeutic drugs. Incorporation of doxorubicin into PLGA nanoparticles allows increasing the antitumor effect of the cytostatics against experimental rat glioblastoma 101.8. Animal survival, tumor volume, and oncogene expression in tumor cells were compared after early (days 2, 5, and 8 after tumor implantation) and late (days 8, 11, and 14) start of the therapy. At late start, a significant increase in the expression of oncogenes Gdnf, Pdgfra, and Melk and genes determining the development of multidrug resistance Abcb1b and Mgmt was revealed. At early start of therapy, only the expression of oncogenes Gdnf, Pdgfra, and Melk was enhanced. Early start of treatment prolonged the survival time and increased tumor growth inhibition by 141.4 and 95.7%, respectively, in comparison with the untreated group; these differences were not observed in the group with late start of therapy. The results indicate that the time of initiation of therapy is a critical parameter affecting the antitumor efficacy of DOX-PLGA.


Asunto(s)
Doxorrubicina , Glioblastoma , Nanopartículas , Animales , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Ratas , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/uso terapéutico , Masculino , Línea Celular Tumoral , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Ácido Poliglicólico/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
3.
Proc Natl Acad Sci U S A ; 121(22): e2314533121, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38776373

RESUMEN

Nanoparticles tethered with vasculature-binding epitopes have been used to deliver the drug into injured or diseased tissues via the bloodstream. However, the extent that blood flow dynamics affects nanoparticle retention at the target site after adhesion needs to be better understood. This knowledge gap potentially underlies significantly different therapeutic efficacies between animal models and humans. An experimentally validated mathematical model that accurately simulates the effects of blood flow on nanoparticle adhesion and retention, thus circumventing the limitations of conventional trial-and-error-based drug design in animal models, is lacking. This paper addresses this technical bottleneck and presents an integrated mathematical method that derives heavily from a unique combination of a mechanics-based dispersion model for nanoparticle transport and diffusion in the boundary layers, an asperity model to account for surface roughness of endothelium, and an experimentally calibrated stochastic nanoparticle-cell adhesion model to describe nanoparticle adhesion and subsequent retention at the target site under external flow. PLGA-b-HA nanoparticles tethered with VHSPNKK peptides that specifically bind to vascular cell adhesion molecules on the inflamed vascular wall were investigated. The computational model revealed that larger particles perform better in adhesion and retention at the endothelium for the particle sizes suitable for drug delivery applications and within physiologically relevant shear rates. The computational model corresponded closely to the in vitro experiments which demonstrates the impact that model-based simulations can have on optimizing nanocarriers in vascular microenvironments, thereby substantially reducing in vivo experimentation as well as the development costs.


Asunto(s)
Nanopartículas , Nanopartículas/química , Humanos , Ligandos , Sistemas de Liberación de Medicamentos/métodos , Adhesión Celular , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
4.
ACS Nano ; 18(20): 13361-13376, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38728619

RESUMEN

Oxygen therapy cannot rescue local lung hypoxia in patients with severe respiratory failure. Here, an inhalable platform is reported for overcoming the aberrant hypoxia-induced immune changes and alveolar damage using camouflaged poly(lactic-co-glycolic) acid (PLGA) microparticles with macrophage apoptotic body membrane (cMAB). cMABs are preloaded with mitochondria-targeting superoxide dismutase/catalase nanocomplexes (NCs) and modified with pathology-responsive macrophage growth factor colony-stimulating factor (CSF) chains, which form a core-shell platform called C-cMAB/NC with efficient deposition in deeper alveoli and high affinity to alveolar epithelial cells (AECs) after CSF chains are cleaved by matrix metalloproteinase 9. Therefore, NCs can be effectively transported into mitochondria to inhibit inflammasome-mediated AECs damage in mouse models of hypoxic acute lung injury. Additionally, the at-site CSF release is sufficient to rescue circulating monocytes and macrophages and alter their phenotypes, maximizing synergetic effects of NCs on creating a pro-regenerative microenvironment that enables resolution of lung injury and inflammation. This inhalable platform may have applications to numerous inflammatory lung diseases.


Asunto(s)
Macrófagos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Animales , Ratones , Macrófagos/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ratones Endogámicos C57BL , Hipoxia , Lesión Pulmonar Aguda/patología , Lesión Pulmonar/patología , Lesión Pulmonar/terapia , Administración por Inhalación , Apoptosis/efectos de los fármacos
5.
Pak J Pharm Sci ; 37(1): 107-113, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38741406

RESUMEN

Entecavir, an effective anti-hepatitis B drug with low resistance rate, was designed as sustained-release micro spheres in our previous study. Here, we aimed to reveal the drug-release mechanism by observing the drug distribution and degradation behavior of poly (lactic-co-glycolic acid) and to investigate the pharmacodynamics of entecavir micro spheres. Raman spectroscopy was used to analyze the distribution of active pharmaceutical ingredients in the micro spheres. The results showed that there was little entecavir near the micro sphere surface. With increasing micro sphere depth, the drug distribution gradually increased and larger-size entecavir crystals were mainly distributed near the spherical center. The degradation behavior of poly (lactic-co-glycolic acid) was investigated using gel permeation chromatography. Changes in poly (lactic-co-glycolic acid) molecular weights during micro sphere degradation revealed that dissolution dominated the release process, which proved our previous research results. Pharmacodynamics studies on transgenic mice indicated that the anti-hepatitis B virus replication effect was maintained for 42 days after a single injection of entecavir micro spheres, similar to the effect of daily oral administration of entecavir tablets for 28 days. The entecavir micro spheres prepared in this study had a good anti-hepatitis B virus replication effect and it is expected to be used in anti hepatitis B virus treatment against hepatitis B virus.


Asunto(s)
Antivirales , Guanina , Virus de la Hepatitis B , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Guanina/farmacología , Guanina/análogos & derivados , Guanina/farmacocinética , Animales , Antivirales/farmacología , Antivirales/farmacocinética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Virus de la Hepatitis B/efectos de los fármacos , Liberación de Fármacos , Ratones Transgénicos , Ratones , Replicación Viral/efectos de los fármacos , Microesferas , Preparaciones de Acción Retardada , Hepatitis B/tratamiento farmacológico , Tamaño de la Partícula , Ácido Poliglicólico/química , Espectrometría Raman , Ácido Láctico
6.
J Nanobiotechnology ; 22(1): 223, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702815

RESUMEN

Cardiac muscle targeting is a notoriously difficult task. Although various nanoparticle (NP) and adeno-associated viral (AAV) strategies with heart tissue tropism have been developed, their performance remains suboptimal. Significant off-target accumulation of i.v.-delivered pharmacotherapies has thwarted development of disease-modifying cardiac treatments, such as gene transfer and gene editing, that may address both rare and highly prevalent cardiomyopathies and their complications. Here, we present an intriguing discovery: cargo-less, safe poly (lactic-co-glycolic acid) particles that drastically improve heart delivery of AAVs and NPs. Our lead formulation is referred to as ePL (enhancer polymer). We show that ePL increases selectivity of AAVs and virus-like NPs (VLNPs) to the heart and de-targets them from the liver. Serotypes known to have high (AAVrh.74) and low (AAV1) heart tissue tropisms were tested with and without ePL. We demonstrate up to an order of magnitude increase in heart-to-liver accumulation ratios in ePL-injected mice. We also show that ePL exhibits AAV/NP-independent mechanisms of action, increasing glucose uptake in the heart, increasing cardiac protein glycosylation, reducing AAV neutralizing antibodies, and delaying blood clearance of AAV/NPs. Current approaches utilizing AAVs or NPs are fraught with challenges related to the low transduction of cardiomyocytes and life-threatening immune responses; our study introduces an exciting possibility to direct these modalities to the heart at reduced i.v. doses and, thus, has an unprecedented impact on drug delivery and gene therapy. Based on our current data, the ePL system is potentially compatible with any therapeutic modality, opening a possibility of cardiac targeting with numerous pharmacological approaches.


Asunto(s)
Dependovirus , Vectores Genéticos , Miocardio , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Dependovirus/genética , Animales , Nanopartículas/química , Ratones , Miocardio/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Humanos , Ratones Endogámicos C57BL , Corazón , Terapia Genética/métodos , Técnicas de Transferencia de Gen , Hígado/metabolismo , Tropismo Viral , Células HEK293
7.
Antiviral Res ; 226: 105900, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38705200

RESUMEN

BACKGROUND & AIMS: The spread of foot-and-mouth disease virus (FMDV) through aerosol droplets among cloven-hoofed ungulates in close contact is a major obstacle for successful animal husbandry. Therefore, the development of suitable mucosal vaccines, especially nasal vaccines, to block the virus at the initial site of infection is crucial. PATIENTS AND METHODS: Here, we constructed eukaryotic expression plasmids containing the T and B-cell epitopes (pTB) of FMDV in tandem with the molecular mucosal adjuvant Fms-like tyrosine kinase receptor 3 ligand (Flt3 ligand, FL) (pTB-FL). Then, the constructed plasmid was electrostatically attached to mannose-modified chitosan-coated poly(lactic-co-glycolic) acid (PLGA) nanospheres (MCS-PLGA-NPs) to obtain an active nasal vaccine targeting the mannose-receptor on the surface of antigen-presenting cells (APCs). RESULTS: The MCS-PLGA-NPs loaded with pTB-FL not only induced a local mucosal immune response, but also induced a systemic immune response in mice. More importantly, the nasal vaccine afforded an 80% protection rate against a highly virulent FMDV strain (AF72) when it was subcutaneously injected into the soles of the feet of guinea pigs. CONCLUSIONS: The nasal vaccine prepared in this study can effectively induce a cross-protective immune response against the challenge with FMDV of same serotype in animals and is promising as a potential FMDV vaccine.


Asunto(s)
Administración Intranasal , Quitosano , Virus de la Fiebre Aftosa , Fiebre Aftosa , Nanosferas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Vacunas Virales , Animales , Quitosano/química , Quitosano/administración & dosificación , Virus de la Fiebre Aftosa/inmunología , Virus de la Fiebre Aftosa/genética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Fiebre Aftosa/prevención & control , Fiebre Aftosa/inmunología , Ratones , Nanosferas/química , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Ratones Endogámicos BALB C , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Femenino , Ácidos Nucleicos/administración & dosificación , Inmunidad Mucosa , Sistemas de Liberación de Medicamentos
8.
Biomater Adv ; 160: 213833, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38564997

RESUMEN

Conventional chemotherapy and poor targeted delivery in brain cancer resulting to poor treatment and develop resistance to anticancer drugs. Meanwhile, it is quite challenging to diagnose/detection of brain tumor at early stage of cancer which resulting in severity of the disease. Despite extensive research, effective treatment with real-time imaging still remains completely unavailable, yet. In this study, two brain cancer cell specific moieties i.e., AS1411 aptamer and RGD are decorated on the surface of chitosan-PLGA nanoparticles to improve targeted co-delivery of docetaxel (DTX) and upconversion nanoparticles (UCNP) for effective brain tumor therapy and real-time imaging. The nanoparticles were developed by a slightly modified emulsion/solvent evaporation method. This investigation also translates the successful synthesis of TPGS-chitosan, TPGS-RGD and TPGS-AS1411 aptamer conjugates for making PLGA nanoparticle as a potential tool of the targeted co-delivery of DTX and UCNP to the brain cancer cells. The developed nanoparticles have shown an average particle size <200 nm, spherical in shape, high encapsulation of DTX and UCNP in the core of nanoparticles, and sustained release of DTX up to 72 h in phosphate buffer saline (pH 7.4). AS1411 aptamer and RGD functionalized theranostic chitosan-PLGA nanoparticles containing DTX and UCNP (DUCPN-RGD-AS1411) have achieved greater cellular uptake, 89-fold improved cytotoxicity, enhanced cancer cell arrest even at lower drug conc., improved bioavailability with higher mean residence time of DTX in systemic circulation and brain tissues. Moreover, DUCPN-RGD-AS1411 have greatly facilitated cellular internalization and higher accumulation of UCNP in brain tissues. Additionally, DUCPN-RGD-AS1411 demonstrated a significant suppression in tumor growth in brain-tumor bearing xenograft BALB/c nude mice with no impressive sign of toxicities. DUCPN-RGD-AS1411 has great potential to be utilized as an effective and safe theranostic tool for brain cancer and other life-threatening cancer therapies.


Asunto(s)
Aptámeros de Nucleótidos , Neoplasias Encefálicas , Quitosano , Docetaxel , Oligodesoxirribonucleótidos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Animales , Humanos , Ratones , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Aptámeros de Nucleótidos/administración & dosificación , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacocinética , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Quitosano/química , Docetaxel/farmacocinética , Docetaxel/administración & dosificación , Docetaxel/farmacología , Docetaxel/uso terapéutico , Nanopartículas/química , Oligopéptidos/química , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacocinética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Nanomedicina Teranóstica/métodos
9.
Nanoscale ; 16(19): 9412-9425, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38650478

RESUMEN

Nanotechnology has the potential to provide formulations of antitumor agents with increased selectivity towards cancer tissue thereby decreasing systemic toxicity. This in vivo study evaluated the potential of novel nanoformulation based on poly(lactic-co-glycolic acid) (PLGA) to reduce the cardiotoxic potential of doxorubicin (DOX). In vivo toxicity of PLGADOX was compared with clinically approved non-PEGylated, liposomal nanoformulation of DOX (LipoDOX) and conventional DOX form (ConvDOX). The study was performed using Wistar Han rats of both sexes that were treated intravenously for 28 days with 5 doses of tested substances at intervals of 5 days. Histopathological analyses of heart tissues showed the presence of myofiber necrosis, degeneration processes, myocytolysis, and hemorrhage after treatment with ConvDOX, whereas only myofiber degeneration and hemorrhage were present after the treatment with nanoformulations. All DOX formulations caused an increase in the troponin T with the greatest increase caused by convDOX. qPCR analyses revealed an increase in the expression of inflammatory markers IL-6 and IL-8 after ConvDOX and an increase in IL-8 expression after lipoDOX treatments. The mass spectra imaging (MSI) of heart tissue indicates numerous metabolic and lipidomic changes caused by ConvDOX, while less severe cardiac damages were found after treatment with nanoformulations. In the case of LipoDOX, autophagy and apoptosis were still detectable, whereas PLGADOX induced only detectable mitochondrial toxicity. Cardiotoxic effects were frequently sex-related with the greater risk of cardiotoxicity observed mostly in male rats.


Asunto(s)
Cardiotoxicidad , Doxorrubicina , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas Wistar , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/análogos & derivados , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ratas , Masculino , Cardiotoxicidad/prevención & control , Femenino , Apoptosis/efectos de los fármacos , Nanopartículas/química , Miocardio/patología , Miocardio/metabolismo , Polietilenglicoles/química , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacología , Corazón/efectos de los fármacos , Liposomas/química
10.
J Mater Chem B ; 12(20): 4935-4944, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38683039

RESUMEN

The oral cavity, a warm and moist environment, is prone to the proliferation of microorganisms like Candida albicans (C. albicans), which forms robust biofilms on biotic and abiotic surfaces, leading to challenging infections. These biofilms are resistant to conventional treatments due to their resilience against antimicrobials and immune responses. The dynamic nature of the oral cavity, including the salivary flow and varying surface properties, complicates the delivery of therapeutic agents. To address these challenges, we introduce dendritic microparticles engineered for enhanced adhesion to dental surfaces and effective delivery of antifungal agents and antibiofilm enzymes. These microparticles are fabricated using a water-in-oil-in-water emulsion process involving a blend of poly(lactic-co-glycolic acid) (PLGA) random copolymer (RCP) and PLGA-b-poly(ethylene glycol) (PLGA-b-PEG) block copolymer (BCP), resulting in particles with surface dendrites that exhibit strong adhesion to oral surfaces. Our study demonstrates the potential of these adhesive microparticles for oral applications. The adhesion tests on various oral surfaces, including dental resin, hydroxyapatite, tooth enamel, and mucosal tissues, reveal superior adhesion of these microparticles compared to conventional spherical ones. Furthermore, the release kinetics of nystatin from these microparticles show a sustained release pattern that can kill C. albicans. The biodegradation of these microparticles on tooth surfaces and their efficacy in preventing fungal biofilms have also been demonstrated. Our findings highlight the effectiveness of adhesive microparticles in delivering therapeutic agents within the oral cavity, offering a promising approach to combat biofilm-associated infections.


Asunto(s)
Antifúngicos , Biopelículas , Candida albicans , Propiedades de Superficie , Biopelículas/efectos de los fármacos , Candida albicans/efectos de los fármacos , Candida albicans/fisiología , Antifúngicos/farmacología , Antifúngicos/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Humanos , Boca/microbiología , Adhesivos/química , Adhesivos/farmacología , Tamaño de la Partícula , Polietilenglicoles/química , Portadores de Fármacos/química
11.
ACS Appl Bio Mater ; 7(5): 3086-3095, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38652779

RESUMEN

Of the 27 million surgeries performed in the United States each year, a reported 2.6% result in a surgical site infection (SSI), and Staphylococci species are commonly the culprit. Alternative therapies, such as nitric oxide (NO)-releasing biomaterials, are being developed to address this issue. NO is a potent antimicrobial agent with several modes of action, including oxidative and nitrosative damage, disruption of bacterial membranes, and dispersion of biofilms. For targeted antibacterial effects, NO is delivered by exogenous donor molecules, like S-nitroso-N-acetylpenicillamine (SNAP). Herein, the impregnation of SNAP into poly(lactic-co-glycolic acid) (PLGA) for SSI prevention is reported for the first time. The NO-releasing PLGA copolymer is fabricated and characterized by donor molecule loading, leaching, and the amount remaining after ethylene oxide sterilization. The swelling ratio, water uptake, static water contact angle, and tensile strength are also investigated. Furthermore, its cytocompatibility is tested against 3T3 mouse fibroblast cells, and its antimicrobial efficacy is assessed against multiple Staphylococci strains. Overall, the NO-releasing PLGA copolymer holds promise as a suture material for eradicating surgical site infections caused by Staphylococci strains. SNAP impregnation affords robust antibacterial properties while maintaining the cytocompatibility and mechanical integrity.


Asunto(s)
Antibacterianos , Materiales Biocompatibles , Ensayo de Materiales , Óxido Nítrico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Infección de la Herida Quirúrgica , Suturas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Infección de la Herida Quirúrgica/prevención & control , Infección de la Herida Quirúrgica/tratamiento farmacológico , Infección de la Herida Quirúrgica/microbiología , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Ratones , Antibacterianos/farmacología , Antibacterianos/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Pruebas de Sensibilidad Microbiana , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/prevención & control , Tamaño de la Partícula , Staphylococcus aureus/efectos de los fármacos , Staphylococcus/efectos de los fármacos
12.
ACS Biomater Sci Eng ; 10(5): 3387-3400, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38656158

RESUMEN

Given the worldwide problem posed by enteric pathogens, the discovery of safe and efficient intestinal adjuvants combined with novel antigen delivery techniques is essential to the design of mucosal vaccines. In this work, we designed poly (lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs) to codeliver all-trans retinoic acid (atRA), novel antigens, and CpG. To address the insolubility of the intestinal adjuvant atRA, we utilized PLGA to encapsulate atRA and form a "nanocapsid" with polydopamine. By leveraging polydopamine, we adsorbed the water-soluble antigens and the TLR9 agonist CpG onto the NPs' surface, resulting in the pathogen-mimicking PLPCa NPs. In this study, the novel fusion protein (HBf), consisting of the Mycobacterium avium subspecies paratuberculosis antigens HBHA, Ag85B, and Bfra, was coloaded onto the NPs. In vitro, PLPCa NPs were shown to promote the activation and maturation of bone marrow-derived dendritic cells. Additionally, we found that PLPCa NPs created an immune-rich microenvironment at the injection site following intramuscular administration. From the results, the PLPCa NPs induced strong IgA levels in the gut in addition to enhancing powerful systemic immune responses. Consequently, significant declines in the bacterial burden and inflammatory score were noted in PLPCa NPs-treated mice. In summary, PLPCa can serve as a novel and safe vaccine delivery platform against gut pathogens, such as paratuberculosis, capable of activating both systemic and intestinal immunity.


Asunto(s)
Nanopartículas , Paratuberculosis , Animales , Nanopartículas/química , Paratuberculosis/inmunología , Paratuberculosis/prevención & control , Ratones , Tretinoina/química , Tretinoina/farmacología , Mycobacterium avium subsp. paratuberculosis/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/química , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Intestinos/inmunología , Intestinos/microbiología , Ratones Endogámicos C57BL , Femenino , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/administración & dosificación , Vacunas Bacterianas/inmunología , Ratones Endogámicos BALB C
13.
ACS Biomater Sci Eng ; 10(5): 3164-3172, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38671385

RESUMEN

Intestinal adhesion is one of the complications that occurs more frequently after abdominal surgery. Postsurgical intestinal adhesion (PIA) can lead to a series of health problems, including abdominal pain, intestinal obstruction, and female infertility. Currently, hydrogels and nanofibrous films as barriers are often used for preventing PIA formation; however, these kinds of materials have their intrinsic disadvantages. Herein, we developed a dual-structure drug delivery patch consisting of poly lactic-co-glycolic acid (PLGA) nanofibers and a chitosan hydrogel (NHP). PLGA nanofibers loaded with deferoxamine mesylate (DFO) were incorporated into the hydrogel; meanwhile, the hydrogel was loaded with anti-inflammatory drug dexamethasone (DXMS). The rapid degradation of the hydrogel facilitated the release of DXMS at the acute inflammatory stage of the early injury and provided effective anti-inflammatory effects for wound sites. Moreover, PLGA composite nanofibers could provide sustained and stable release of DFO for promoting the peritoneal repair by the angiogenesis effects of DFO. The in vivo results indicated that NHP can effectively prevent PIA formation by restraining inflammation and vascularization, promoting peritoneal repair. Therefore, we believe that our NHP has a great potential application in inhibition of PIA.


Asunto(s)
Dexametasona , Sistemas de Liberación de Medicamentos , Hidrogeles , Nanofibras , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Nanofibras/química , Nanofibras/uso terapéutico , Hidrogeles/química , Hidrogeles/farmacología , Hidrogeles/administración & dosificación , Adherencias Tisulares/prevención & control , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Dexametasona/farmacología , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Quitosano/química , Quitosano/farmacología , Intestinos/efectos de los fármacos , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Complicaciones Posoperatorias/prevención & control , Ratas Sprague-Dawley , Ratones , Femenino , Ratas
14.
Biomater Sci ; 12(10): 2660-2671, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38592706

RESUMEN

The endo-lysosomal pathway is a major barrier for the trans-epithelial transport of nanoparticles (NPs), but escape strategies could facilitate trans-epithelial delivery. Based on the polarization properties of the epithelium, different escape compartments may result in different exocytosis fates of NPs and further affect the delivery efficiency. Therefore, optimizing the escape sites is critical for trans-epithelial delivery. Here, commonly used PEG-coated-poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles were fabricated as model nanoparticles (MNPs) and the intestinal epithelium was chosen as the polarized epithelium. The MNPs were incubated with different endosomolytic agents for early endosomal escape, late endosomal escape and lysosomal escape, respectively. According to in vitro and in vivo studies, MNPs escaping from early endosomes and late endosomes exhibited stronger capacity for trans-epithelial transport than those escaping from lysosomes. By further probing into the mechanism, we surprisingly found that although MNPs escaping from early endosomes quickly egressed from the apical side of epithelia, they were subsequently followed by "reuptake" via caveolae and trafficked through the endoplasmic reticulum-Golgi apparatus (ER/GA) secretory pathway, achieving efficient trans-epithelial transport; MNPs escaping from late endosomes, which were located near the nucleus, were prone to enter the ER/GA for efficient basolateral exocytosis. However, MNPs escaping from lysosomes were detained within cells by autophagosomes. Collectively, our research suggested that early endosomes and late endosomes were ideal escape sites for trans-epithelial delivery.


Asunto(s)
Endosomas , Exocitosis , Lisosomas , Nanopartículas , Lisosomas/metabolismo , Exocitosis/fisiología , Animales , Nanopartículas/química , Endosomas/metabolismo , Polietilenglicoles/química , Humanos , Ratones , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Perros , Mucosa Intestinal/metabolismo
15.
Biomater Sci ; 12(10): 2672-2688, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38596867

RESUMEN

Breast cancer, a pervasive malignancy affecting women, demands a diverse treatment approach including chemotherapy, radiotherapy, and surgical interventions. However, the effectiveness of doxorubicin (DOX), a cornerstone in breast cancer therapy, is limited when used as a monotherapy, and concerns about cardiotoxicity persist. Ginsenoside Rg3, a classic compound of traditional Chinese medicine found in Panax ginseng C. A. Mey., possesses diverse pharmacological properties, including cardiovascular protection, immune modulation, and anticancer effects. Ginsenoside Rg3 is considered a promising candidate for enhancing cancer treatment when combined with chemotherapy agents. Nevertheless, the intrinsic challenges of Rg3, such as its poor water solubility and low oral bioavailability, necessitate innovative solutions. Herein, we developed Rg3-PLGA@TMVs by encapsulating Rg3 within PLGA nanoparticles (Rg3-PLGA) and coating them with membranes derived from tumor cell-derived microvesicles (TMVs). Rg3-PLGA@TMVs displayed an array of favorable advantages, including controlled release, prolonged storage stability, high drug loading efficiency and a remarkable ability to activate dendritic cells in vitro. This activation is evident through the augmentation of CD86+CD80+ dendritic cells, along with a reduction in phagocytic activity and acid phosphatase levels. When combined with DOX, the synergistic effect of Rg3-PLGA@TMVs significantly inhibits 4T1 tumor growth and fosters the development of antitumor immunity in tumor-bearing mice. Most notably, this delivery system effectively mitigates the toxic side effects of DOX, particularly those affecting the heart. Overall, Rg3-PLGA@TMVs provide a novel strategy to enhance the efficacy of DOX while simultaneously mitigating its associated toxicities and demonstrate promising potential for the combined chemo-immunotherapy of breast cancer.


Asunto(s)
Doxorrubicina , Ginsenósidos , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ginsenósidos/química , Ginsenósidos/farmacología , Ginsenósidos/administración & dosificación , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Femenino , Nanopartículas/química , Ratones , Doxorrubicina/farmacología , Doxorrubicina/química , Doxorrubicina/administración & dosificación , Humanos , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/efectos de los fármacos , Ratones Endogámicos BALB C , Línea Celular Tumoral , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Liberación de Fármacos , Portadores de Fármacos/química , Células Dendríticas/efectos de los fármacos
16.
Acta Biomater ; 180: 323-336, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38561075

RESUMEN

Peripheral nerve injuries (PNIs) can cause neuropathies and significantly affect the patient's quality of life. Autograft transplantation is the gold standard for conventional treatment; however, its application is limited by nerve unavailability, size mismatch, and local tissue adhesion. Tissue engineering, such as nerve guidance conduits, is an alternative and promising strategy to guide nerve regeneration for peripheral nerve repair; however, only a few conduits could reach the high repair efficiency of autografts. The healing process of PNI is frequently accompanied by not only axonal and myelination regeneration but also angiogenesis, which initializes nerve regeneration through vascular endothelial growth factor A (VEGF-A). In this study, a composite nerve conduit with a poly (lactic-co-glycolic acid) (PLGA) hollow tube as the outer layer and gelatin methacryloyl (GelMA) encapsulated with VEGF-A transfected Schwann cells (SCs) as the inner layer was established to evaluate its promising ability for peripheral nerve repair. A rat model of peripheral nerve defect was used to examine the efficiency of PLGA/GelMA-SC (VA) conduits, whereas autograft, PLGA, PLGA/GelMA, and PLGA/GelMA-SC (NC) were used as controls. VEGF-A-transfected SCs can provide a stable source for VEGF-A secretion. Furthermore, encapsulation in GelMA cannot only promote proliferation and tube formation of human umbilical vein endothelial cells but also enhance dorsal root ganglia and neuronal cell extension. Previous animal studies have demonstrated that the regenerative effects of PLGA/GelMA-SC (VA) nerve conduit were similar to those of autografts and much better than those of other conduits. These findings indicate that combination of VEGF-A-overexpressing SCs and PLGA/GelMA conduit-guided peripheral nerve repair provides a promising method that enhances angiogenesis and regeneration during nerve repair. STATEMENT OF SIGNIFICANCE: Nerve guidance conduits shows promise for peripheral nerve repair, while achieving the repair efficiency of autografts remains a challenge. In this study, a composite nerve conduit with a PLGA hollow tube as the outer layer and gelatin methacryloyl (GelMA) encapsulated with vascular endothelial growth factor A (VEGF-A)-transfected Schwann cells (SCs) as the inner layer was established to evaluate its potential ability for peripheral nerve repair. This approach preserves growth factor bioactivity and enhances material properties. GelMA insertion promotes Schwann cell proliferation and morphology extension. Moreover, transfected SCs serve as a stable VEGF-A source and fostering angiogenesis. This study offers a method preserving growth factor efficacy and safeguarding SCs, providing a comprehensive solution for enhanced angiogenesis and nerve regeneration.


Asunto(s)
Neovascularización Fisiológica , Regeneración Nerviosa , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas Sprague-Dawley , Células de Schwann , Factor A de Crecimiento Endotelial Vascular , Células de Schwann/metabolismo , Células de Schwann/citología , Animales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Regeneración Nerviosa/efectos de los fármacos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Transfección , Gelatina/química , Masculino , Andamios del Tejido/química , Humanos , Traumatismos de los Nervios Periféricos/terapia , Traumatismos de los Nervios Periféricos/patología , Angiogénesis
17.
Pathol Res Pract ; 257: 155295, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38603841

RESUMEN

Tobacco smoking is a leading cause of preventable mortality, and it is the major contributor to diseases such as COPD and lung cancer. Cigarette smoke compromises the pulmonary antiviral immune response, increasing susceptibility to viral infections. There is currently no therapy that specifically addresses the problem of impaired antiviral response in cigarette smokers and COPD patients, highlighting the necessity to develop novel treatment strategies. 18-ß-glycyrrhetinic acid (18-ß-gly) is a phytoceutical derived from licorice with promising anti-inflammatory, antioxidant, and antiviral activities whose clinical application is hampered by poor solubility. This study explores the therapeutic potential of an advanced drug delivery system encapsulating 18-ß-gly in poly lactic-co-glycolic acid (PLGA) nanoparticles in addressing the impaired antiviral immunity observed in smokers and COPD patients. Exposure of BCi-NS1.1 human bronchial epithelial cells to cigarette smoke extract (CSE) resulted in reduced expression of critical antiviral chemokines (IP-10, I-TAC, MIP-1α/1ß), mimicking what happens in smokers and COPD patients. Treatment with 18-ß-gly-PLGA nanoparticles partially restored the expression of these chemokines, demonstrating promising therapeutic impact. The nanoparticles increased IP-10, I-TAC, and MIP-1α/1ß levels, exhibiting potential in attenuating the negative effects of cigarette smoke on the antiviral response. This study provides a novel approach to address the impaired antiviral immune response in vulnerable populations, offering a foundation for further investigations and potential therapeutic interventions. Further studies, including a comprehensive in vitro characterization and in vivo testing, are warranted to validate the therapeutic efficacy of 18-ß-gly-PLGA nanoparticles in respiratory disorders associated with compromised antiviral immunity.


Asunto(s)
Ácido Glicirretínico , Nanopartículas , Humanos , Ácido Glicirretínico/farmacología , Ácido Glicirretínico/análogos & derivados , Antivirales/farmacología , Humo/efectos adversos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Línea Celular , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Fumar Cigarrillos/efectos adversos
18.
Talanta ; 274: 126005, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38599116

RESUMEN

In preparing monoclonal antibodies by hybridoma cell technology, the quality of B lymphocytes used for cell fusion directly affects the sensitivity of monoclonal antibodies. To obtain B-lymphocytes producing high-quality specific antibodies for cell fusion during the immunization phase of the antigen, we prepared a TH2-Cell stimulatory delivery system as a novel adjuvant. Astragalus polysaccharide has a good ability to enhance antigenic immune response, and it was encapsulated in biocompatible materials PLGA as an immunostimulatory factor to form the delivery system (APS-PLGA). The preparation conditions of APSP were optimized using RSM to attain the highest utilization of APS. Immunization against ZEN-BSA antigen using APSP as an adjuvant to obtain B lymphocytes producing ZEN-specific antibodies for cell fusion. As results present, APSP could induce a stronger TH2 immune response through differentiating CD4 T cells and promoting IL-4 and IL-6 cytokines. Moreover, it could slow down the release efficiency of ZEN-BSA and enhance the targeting of ZEN-BSA to lymph nodes in vivo experiments. Ultimately, the sensitivity of mouse serum ZEN-specific antibodies was enhanced upon completion of immunization, indicating a significant upregulation of high-quality B lymphocyte expression. In the preparation of monoclonal antibodies, the proportion of positive wells for the first screening was 60%, and the inhibition rates of the antibodies were all similar (>50%). Then we obtained the ZEN monoclonal antibody with IC50 of 0.049 ng/mL, which was more sensitive than most antibodies prepared under conventional adjuvants. Finally, a TRFIAS strip assay was preliminarily established with a LOD value of 0.246 ng/mL.


Asunto(s)
Adyuvantes Inmunológicos , Anticuerpos Monoclonales , Linfocitos B , Ratones Endogámicos BALB C , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Linfocitos B/inmunología , Linfocitos B/efectos de los fármacos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/química , Nanopartículas/química , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Ratones , Femenino , Activación de Linfocitos/efectos de los fármacos , Inmunización
19.
Mol Pharm ; 21(5): 2238-2249, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38622497

RESUMEN

Tuberculosis (TB) is a chronic disease caused byMycobacterium tuberculosis (Mtb), which shows a long treatment cycle often leads to drug resistance, making treatment more difficult. Immunogens present in the pathogen's cell membrane can stimulate endogenous immune responses. Therefore, an effective lipid-based vaccine or drug delivery vehicle formulated from the pathogen's cell membrane can improve treatment outcomes. Herein, we extracted and characterized lipids fromMycobacterium smegmatis, and the extracts contained lipids belonging to numerous lipid classes and compounds typically found associated with mycobacteria. The extracted lipids were used to formulate biomimetic lipid reconstituted nanoparticles (LrNs) and LrNs-coated poly(lactic-co-glycolic acid) nanoparticles (PLGA-LrNs). Physiochemical characterization and results of morphology suggested that PLGA-LrNs exhibited enhanced stability compared with LrNs. And both of these two types of nanoparticles inhibited the growth of M. smegmatis. After loading different drugs, PLGA-LrNs containing berberine or coptisine strongly and synergistically prevented the growth of M. smegmatis. Altogether, the bacterial membrane lipids we extracted with antibacterial activity can be used as nanocarrier coating for synergistic antibacterial treatment of M. smegmatis─an alternative model of Mtb, which is expected as a novel therapeutic system for TB treatment.


Asunto(s)
Mycobacterium smegmatis , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Nanopartículas/química , Mycobacterium smegmatis/efectos de los fármacos , Lípidos/química , Sinergismo Farmacológico , Membrana Celular/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/efectos de los fármacos , Antituberculosos/farmacología , Antituberculosos/química , Antituberculosos/administración & dosificación , Mycobacterium/efectos de los fármacos , Berberina/farmacología , Berberina/química , Portadores de Fármacos/química , Tuberculosis/tratamiento farmacológico
20.
Int J Biol Macromol ; 267(Pt 1): 131386, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582458

RESUMEN

Verteporfin (VER), a photosensitizer used in macular degeneration therapy, has shown promise in controlling macrophage polarization and alleviating inflammation in acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). However, its hydrophobicity, limited bioavailability, and side effects hinder its therapeutic potential. In this study, we aimed to enhance the therapeutic potential of VER through pulmonary nebulized drug delivery for ALI/ARDS treatment. We combined hydrophilic hyaluronic acid (HA) with an oil-in-water system containing a poly(lactic acid-co-glycolic acid) (PLGA) copolymer of VER to synthesize HA@PLGA-VER (PHV) nanoparticles with favorable surface characteristics to improve the bioavailability and targeting ability of VER. PHV possesses suitable electrical properties, a narrow size distribution (approximately 200 nm), and favorable stability. In vitro and in vivo studies demonstrated the excellent biocompatibility, safety, and anti-inflammatory responses of the PHV by suppressing M1 macrophage polarization while inducing M2 polarization. The in vivo experiments indicated that the treatment with aerosolized nano-VER (PHV) allowed more drugs to accumulate and penetrate into the lungs, improved the pulmonary function and attenuated lung injury, and mortality of ALI mice, achieving improved therapeutic outcomes. These findings highlight the potential of PHV as a promising delivery system via nebulization for enhancing the therapeutic effects of VER in ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Portadores de Fármacos , Ácido Hialurónico , Nanopartículas , Verteporfina , Lesión Pulmonar Aguda/tratamiento farmacológico , Ácido Hialurónico/química , Animales , Ratones , Verteporfina/administración & dosificación , Verteporfina/farmacología , Verteporfina/uso terapéutico , Nanopartículas/química , Portadores de Fármacos/química , Células RAW 264.7 , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Aerosoles , Masculino , Sistemas de Liberación de Medicamentos , Administración por Inhalación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA